1
|
Tang M, Wu Y, Olnood CG, Gao Y, Wang F, Zhang Z, Peng C, Zhou X, Huang C, Xiong X, Yin Y. Effects of peroxidized lipids on intestinal morphology, antioxidant capacity and gut microbiome in piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 20:430-443. [PMID: 40034456 PMCID: PMC11875184 DOI: 10.1016/j.aninu.2024.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/05/2024] [Accepted: 11/20/2024] [Indexed: 03/05/2025]
Abstract
This study investigated the effect of peroxidized lipids on piglets' growth performance, intestinal morphology, inflammatory reactions, oxidative stress in the liver, duodenum, jejunum, ileum, and colon, and ileal microbiota. Twenty piglets (Duroc × [Landrace × Yorkshire]; age = 21 d old, BW = 6.5 ± 1 kg) were randomly assigned to two groups with 10 replicates per group and one piglet per replicate. The control group was fed 6% fresh soybean oil and the peroxidized soybean oil (PSO) group fed 6% PSO. The experimental feeding period lasted 24 d. The study found no impact on ADFI, ADG and gain to feed ratio (P > 0.05). However, the PSO group increased the diarrhea index and the serum levels of lactate dehydrogenase triglycerides, cholesterol, low-density lipoprotein cholesterol, and high-density lipoprotein cholesterol (P < 0.05), along with decreased concentrations of alanine aminotransferase and blood urea nitrogen (P < 0.05). For oxidative enzymes, PSO increased the concentration of F2-isoprostane in urine (P = 0.032), malondialdehyde (MDA) in the duodenum (P = 0.001) and jejunum (P = 0.004), decreased thiobarbituric acid reactive substances (TBARS) in the liver (P = 0.001) but increased TBARS in duodenum (P = 0.001), and carbonylated proteins in the duodenum (P = 0.003). For antioxidant enzymes, PSO decreased superoxide dismutase (SOD) in the liver (P = 0.001), colon (P = 0.002), and jejunum (P = 0.015), along with glutathione peroxidase (GSH-Px) in the liver (P = 0.008) and NAD(P)H:quinone oxidoreductase 1 (NQO1) in ileum (P = 0.001). For inflammatory reactions, PSO increased interleukin (IL)-1β concentrations in the duodenum and colon, and IL-10 in the jejunum, while decreasing IL-4 concentration in the duodenum (P < 0.05). For intestinal morphology and ileal microbiota, PSO increased ileal crypt depth, while decreasing the crypt-to-villus ratio (P < 0.05). Peroxidized soybean oil increased the relative abundance of Prevotella, Clostridium_sensu_stricto_1, Clostridium_sensu_stricto_6, Pasteurella and Klebsiella (P < 0.05). In conclusion, this study revealed that PSO worsened diarrhea, increasing the ileal crypt depth and the relative abundance of harmful microbiota, and induced oxidative stress and inflammation in the intestines and liver, primarily in the jejunum and ileum.
Collapse
Affiliation(s)
- Mengxuan Tang
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Yuliang Wu
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
| | | | - Yundi Gao
- Sichuan Synlight Biotech Ltd., Chengdu 610041, China
| | - Fei Wang
- Sichuan Synlight Biotech Ltd., Chengdu 610041, China
| | - Zicheng Zhang
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Can Peng
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
| | - Xihong Zhou
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
| | - Chunxia Huang
- School of Stomatology, Changsha Medical University, Changsha 410219, China
| | - Xia Xiong
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
- School of Stomatology, Changsha Medical University, Changsha 410219, China
| | - Yulong Yin
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
| |
Collapse
|
2
|
Ibrahim H, Sharawy MH, Hamed MF, Abu-Elsaad N. Peficitinib halts acute kidney injury via JAK/STAT3 and growth factors immunomodulation. Eur J Pharmacol 2024; 984:177020. [PMID: 39349115 DOI: 10.1016/j.ejphar.2024.177020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/27/2024] [Accepted: 09/26/2024] [Indexed: 10/02/2024]
Abstract
Acute Kidney Injury (AKI) is characterized by a sudden loss of kidney function and its management continues to be a challenge. In this study the effect of peficitinib, a Janus kinase inhibitor (JAKi), was studied in an aim to stop the progression of AKI at an early point of injury. Adult male mice were injected with aristolochic acid (AA) a single dose (10 mg/kg, i.p) to induce AKI. Peficitinib was injected in one of the two tested doses (5 or 10 mg/kg, i.p) 1 h after AA injection and was continued daily for seven days. Histopathological evaluation showed that peficitinib alleviated necrosis and hyaline cast formation induced by aristolochic acid. It decreased serum creatinine and the kidney injury molecule-1 (KIM-1) elevated by AA. Peficitinib also mitigated AA induced oxidative stress through regulating total antioxidant capacity (TAC) and reduced glutathione (GSH) level in renal tissue. Additionally, renal sections isolated from groups that received peficitinib revealed a decrease in vascular endothelial growth factor receptor 1 interstitial expression and transforming growth factor-beta 1 (TGF-β1) renal level. Peficitinib received groups showed a decrease in the active phosphorylated form of signal transducers and activators of transcription (STAT3). Moreover, peficitinib decreased renal protein levels and gene expression of the pro-inflammatory cytokines; interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α) and interferon gamma (IFN-γ). These findings suggest that peficitinib is helpful in halting AKI progression into chronic kidney disease through modulating JAK/STAT3 dependent inflammatory pathways and growth factors involved in normal glomerular function.
Collapse
Affiliation(s)
- Hassnaa Ibrahim
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Mansoura, 33516, Egypt; Pharmacist at Urology and Nephrology Center, Mansoura University, Mansoura, 33516, Egypt
| | - Maha H Sharawy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Mansoura, 33516, Egypt.
| | - Mohamed F Hamed
- Pathology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 33516, Egypt
| | - Nashwa Abu-Elsaad
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Mansoura, 33516, Egypt
| |
Collapse
|
3
|
Ma Y, Du C, Liu Y, Feng M, Shou Y, Yu D, Jin Y. Aristolochic acid-induced dyslipidemia and hepatotoxicity: The potential role of FXR and AHR receptors. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 287:117266. [PMID: 39509784 DOI: 10.1016/j.ecoenv.2024.117266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/26/2024] [Accepted: 10/27/2024] [Indexed: 11/15/2024]
Abstract
Aristolochic acids (AAs) represent a class of nitrophenanthrene carboxylic acids naturally existing or accidentally mixed in herbal medicines or crops, which have long been recognized for causing nephropathy. Recently, the linkage between AAs and liver injury has become a concern; however, the current understanding of the mechanism or mode of action (MOA) is limited. In the present study, we investigated nuclear receptor-mediated MOA associated with AAs-induced liver injury including dyslipidemia and hepatotoxicity. Bioinformatic analysis of AAI-interacting genes indicated nuclear receptor-mediated metabolizing pathways; Transcriptomic profiling of AAs-exposed rats with liver injury suggested FXR-, NRF2-, and AHR- mediated pathways in the injured livers of the rats. Mechanistic investigation using HepG2 cells indicated AAI-induced hepatic lipid accumulation by elevating Triglyceride (TG) through inhibition of the FXR. In addition, AAI-induced hepatocellular damage by activating the AHR pathway, which further generated ROS and activated the NRF2 pathway. Together, these results provided new clues for researchers who are interested in chemical-induced liver injury.
Collapse
Affiliation(s)
- Yumei Ma
- School of Public Health, Qingdao University, Qingdao, China
| | - Chenlong Du
- Ningxia Hui Autonomous Region Center for Disease Control and Prevention, Yinchuan, China
| | - Yuzhen Liu
- Gaomi Municipal Center for Disease Control and Prevention, Weifang Institute of Preventive Medicine, Weifang, China
| | - Meiyao Feng
- Department of Environmental Health, Qingdao Municipal Center for Disease Control and Prevention, Qingdao Institute of Preventive Medicine, Qingdao, China
| | - Yingqing Shou
- School of Public Health, Qingdao University, Qingdao, China
| | - Dianke Yu
- School of Public Health, Qingdao University, Qingdao, China
| | - Yuan Jin
- School of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
4
|
Marin DE, Bulgaru VC, Pertea A, Grosu IA, Pistol GC, Taranu I. Alternariol Monomethyl-Ether Induces Toxicity via Cell Death and Oxidative Stress in Swine Intestinal Epithelial Cells. Toxins (Basel) 2024; 16:223. [PMID: 38787075 PMCID: PMC11125839 DOI: 10.3390/toxins16050223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/22/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Alternariol monomethyl-ether (AME), together with altenuene and alternariol, belongs to the Alternaria mycotoxins group, which can contaminate different substrates, including cereals. The aim of the present study was to obtain a deeper understanding concerning the effects of AME on pig intestinal health using epithelial intestinal cell lines as the data concerning the possible effects of Alternaria toxins on swine are scarce and insufficient for assessing the risk represented by Alternaria toxins for animal health. Our results have shown a dose-related effect on IPEC-1 cell viability, with an IC50 value of 10.5 μM. Exposure to the toxin induced an increase in total apoptotic cells, suggesting that AME induces programmed cell death through apoptosis based on caspase-3/7 activation in IPEC-1 cells. DNA and protein oxidative damage triggered by AME were associated with an alteration of the antioxidant response, as shown by a decrease in the enzymatic activity of catalase and superoxide dismutase. These effects on the oxidative response can be related to an inhibition of the Akt/Nrf2/HO-1 signaling pathway; however, further studies are needed in order to validate these in vitro data using in vivo trials in swine.
Collapse
Affiliation(s)
- Daniela Eliza Marin
- National Research and Development Institute for Biology and Animal Nutrition (INCDBNA-IBNA-Balotesti), Calea Bucuresti nr.1, 077015 Balotesti Ilfov, Romania; (V.C.B.); (A.P.); (I.A.G.); (G.C.P.); (I.T.)
| | | | | | | | | | | |
Collapse
|
5
|
Hong C, Huang Y, Cao S, Wang L, Yang X, Hu S, Gao K, Jiang Z, Xiao H. Accurate models and nutritional strategies for specific oxidative stress factors: Does the dose matter in swine production? J Anim Sci Biotechnol 2024; 15:11. [PMID: 38273345 PMCID: PMC10811888 DOI: 10.1186/s40104-023-00964-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/01/2023] [Indexed: 01/27/2024] Open
Abstract
Oxidative stress has been associated with a number of physiological problems in swine, including reduced production efficiency. Recently, although there has been increased research into regulatory mechanisms and antioxidant strategies in relation to oxidative stress-induced pig production, it remains so far largely unsuccessful to develop accurate models and nutritional strategies for specific oxidative stress factors. Here, we discuss the dose and dose intensity of the causes of oxidative stress involving physiological, environmental and dietary factors, recent research models and the antioxidant strategies to provide theoretical guidance for future oxidative stress research in swine.
Collapse
Affiliation(s)
- Changming Hong
- State Key Laboratory of Swine and Poultry Breeding Industry, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Yujian Huang
- State Key Laboratory of Swine and Poultry Breeding Industry, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Shuting Cao
- State Key Laboratory of Swine and Poultry Breeding Industry, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Li Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Xuefen Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Shenglan Hu
- State Key Laboratory of Swine and Poultry Breeding Industry, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Kaiguo Gao
- State Key Laboratory of Swine and Poultry Breeding Industry, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Zongyong Jiang
- State Key Laboratory of Swine and Poultry Breeding Industry, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Hao Xiao
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, 1 Dafeng 1st Street, Guangzhou, 510640, China.
| |
Collapse
|
6
|
EFSA Panel on Contaminants in the Food Chain (CONTAM), Schrenk D, Bignami M, Bodin L, Chipman JK, del Mazo J, Grasl‐Kraupp B, Hogstrand C, Hoogenboom L(R, Leblanc J, Nielsen E, Ntzani E, Sand S, Schwerdtle T, Vleminckx C, Wallace H, Gropp J, Antonissen G, Rychen G, Gómez Ruiz JÁ, Innocenti ML, Rovesti E, Petersen A. Risks for animal health related to the presence of ochratoxin A (OTA) in feed. EFSA J 2023; 21:e08375. [PMID: 37942224 PMCID: PMC10628734 DOI: 10.2903/j.efsa.2023.8375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023] Open
Abstract
In 2004, the EFSA Panel on Contaminants in the Food Chain (CONTAM) adopted a Scientific Opinion on the risks to animal health and transfer from feed to food of animal origin related to the presence of ochratoxin A (OTA) in feed. The European Commission requested EFSA to assess newly available scientific information and to update the 2004 Scientific Opinion. OTA is produced by several fungi of the genera Aspergillus and Penicillium. In most animal species it is rapidly and extensively absorbed in the gastro-intestinal tract, binds strongly to plasma albumins and is mainly detoxified to ochratoxin alpha (OTalpha) by ruminal microbiota. In pigs, OTA has been found mainly in liver and kidney. Transfer of OTA from feed to milk in ruminants and donkeys as well as to eggs from poultry is confirmed but low. Overall, OTA impairs function and structure of kidneys and liver, causes immunosuppression and affects the zootechnical performance (e.g. body weight gain, feed/gain ratio, etc.), with monogastric species being more susceptible than ruminants because of limited detoxification to OTalpha. The CONTAM Panel considered as reference point (RP) for adverse animal health effects: for pigs and rabbits 0.01 mg OTA/kg feed, for chickens for fattening and hens 0.03 mg OTA/kg feed. A total of 9,184 analytical results on OTA in feed, expressed in dry matter, were available. Dietary exposure was assessed using different scenarios based on either model diets or compound feed (complete feed or complementary feed plus forage). Risk characterisation was made for the animals for which an RP could be identified. The CONTAM Panel considers that the risk related to OTA in feed for adverse health effects for pigs, chickens for fattening, hens and rabbits is low.
Collapse
|
7
|
Popescu RG, Marinescu GC, Rădulescu AL, Marin DE, Țăranu I, Dinischiotu A. Natural Antioxidant By-Product Mixture Counteracts the Effects of Aflatoxin B1 and Ochratoxin A Exposure of Piglets after Weaning: A Proteomic Survey on Liver Microsomal Fraction. Toxins (Basel) 2023; 15:toxins15040299. [PMID: 37104237 PMCID: PMC10143337 DOI: 10.3390/toxins15040299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/28/2023] Open
Abstract
Mycotoxins are toxic compounds produced by certain strains of fungi that can contaminate raw feed materials. Once ingested, even in small doses, they cause multiple health issues for animals and, downstream, for people consuming meat. It was proposed that inclusion of antioxidant-rich plant-derived feed might diminish the harmful effects of mycotoxins, maintaining the farm animals' health and meat quality for human consumption. This work investigates the large scale proteomic effects on piglets' liver of aflatoxin B1 and ochratoxin A mycotoxins and the potential compensatory effects of grapeseed and sea buckthorn meal administration as dietary byproduct antioxidants against mycotoxins' damage. Forty cross-bred TOPIGS-40 hybrid piglets after weaning were assigned to three (n = 10) experimental groups (A, M, AM) and one control group (C) and fed with experimental diets for 30 days. After 4 weeks, liver samples were collected, and the microsomal fraction was isolated. Unbiased label-free, library-free, data-independent acquisition (DIA) mass spectrometry SWATH methods were able to relatively quantify 1878 proteins from piglets' liver microsomes, confirming previously reported effects on metabolism of xenobiotics by cytochrome P450, TCA cycle, glutathione synthesis and use, and oxidative phosphorylation. Pathways enrichment revealed that fatty acid metabolism, steroid biosynthesis, regulation of actin cytoskeleton, regulation of gene expression by spliceosomes, membrane trafficking, peroxisome, thermogenesis, retinol, pyruvate, and amino acids metabolism pathways are also affected by the mycotoxins. Antioxidants restored expression level of proteins PRDX3, AGL, PYGL, fatty acids biosynthesis, endoplasmic reticulum, peroxisome, amino acid synthesis pathways, and, partially, OXPHOS mitochondrial subunits. However, excess of antioxidants might cause significant changes in CYP2C301, PPP4R4, COL18A1, UBASH3A, and other proteins expression levels. Future analysis of proteomics data corelated to animals growing performance and meat quality studies are necessary.
Collapse
Affiliation(s)
- Roua Gabriela Popescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Splaiul Independentei No. 91-95, 050095 Bucharest, Romania
- Independent Research Association, Timisului No. 58, 012416 Bucharest, Romania
| | - George Cătălin Marinescu
- Independent Research Association, Timisului No. 58, 012416 Bucharest, Romania
- Blue Screen SRL, Timisului No. 58, 012416 Bucharest, Romania
| | - Andreea Luminița Rădulescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Splaiul Independentei No. 91-95, 050095 Bucharest, Romania
| | - Daniela Eliza Marin
- Laboratory of Animal Biology, National Institute for Research and Development for Biology and Animal Nutrition, Calea Bucuresti No. 1, 077015 Balotesti, Romania
| | - Ionelia Țăranu
- Laboratory of Animal Biology, National Institute for Research and Development for Biology and Animal Nutrition, Calea Bucuresti No. 1, 077015 Balotesti, Romania
| | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Splaiul Independentei No. 91-95, 050095 Bucharest, Romania
| |
Collapse
|
8
|
Effect of Dioscorea Opposite Waste Supplementation on Antioxidant Capacity, Immune Response and Rumen Microbiome in Weaned Lambs. FERMENTATION-BASEL 2023. [DOI: 10.3390/fermentation9030256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Dioscorea opposite waste (DOW) has been shown to improve the gastrointestinal microbiome, antioxidation capacity, and immune activity, indicating it is a potential feed resource to improve the physiological health and rumen function of weaned lambs. In the present study, the responses of rumen microbiome to DOW supplementation in diet were profiled using metagenome sequencing. In addition, the potential of DOW to regulate plasma parameters in weaned lambs and its possible mechanisms were investigated. Sixty healthy male small tail Han lambs (22.68 ± 2.56 kg) were selected and equally assigned to four dietary treatments: (1) DOW-free diet (CON), (2) addition of 10% DOW diet (DOW1), (3) addition of 15% DOW diet (DOW2), and (4) addition of 20% DOW diet (DOW3). Experimental lambs were fed a corresponding diet for 62 days. Rumen microbiome and plasma parameters were determined at the end of the experiment. The results showed that dietary supplementation with DOW linearly increased the concentration of aspartate aminotransferase, alkaline phosphatase, Immunoglobulin A, Immunoglobulin M, Immunoglobulin G, Glutathione peroxidase, Superoxide dismutase, and total antioxidant capacity in the plasma of weaned lambs, but an opposite trend was observed in Interleukin-1β, Interleukin-6, tumor necrosis factor-α, and Malondialdehyde between the DOW-supplemented group and the CON group. Sequencing of rumen metagenome revealed that dietary supplementation with 20% DOW significantly affected the microbial composition and function and increased the richness and diversity of rumen microbiota and relative abundance of phylum Verrucomicrobia, Planctomycetes, Fibrobacteres, Chloroflexi, Actinobacteria, and Acidobacteria and species Ruminococcaceae_bacterium, Clostridiales_bacterium_NK3B98, Clostridiales_bacterium, and Clostridia_bacterium. It was concluded that supplementing the weaned lamb’s ration with DOW increased the immune response and antioxidant capacity in a dose-dependent manner. Meanwhile, dietary supplementation with 20% DOW modulated the composition of rumen microbiome function by increasing Ruminococcaceae_bacterium and Clostridiales_bacterium with improving the polysaccharide hydrolase activity in the rumen.
Collapse
|
9
|
Zhang Z, Sun Y, Xie H, Wang J, Zhang X, Shi Z, Liu Y. Protective effect of selenomethionine on kidney injury induced by ochratoxin A in rabbits. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:29874-29887. [PMID: 36417076 DOI: 10.1007/s11356-022-24297-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/14/2022] [Indexed: 06/16/2023]
Abstract
The purpose of this study was to investigate the protective effect and mechanism of selenomethionine (SeMet) on ochratoxin A (OTA)-induced nephrotoxicity in rabbits. In total, sixty Ira rabbits were randomly divided into 5 groups (the control group, OTA group, 0. 2 mg/kg SeMet + OTA group, 0. 4 mg/kg SeMet + OTA group, and 0. 6 mg/kg SeMet + OTA group). The rabbits were fed diets supplemented with different doses of SeMet for 21 days and given 0. 2 mg/kg OTA starting on day 15 for a week. The results showed that the SeMet supplementation could improve the changes in blood physiological indices and renal function decline caused by OTA poisoning, and alleviate pathological kidney injury in the rabbits. SeMet also increased the activities of total antioxidant capacity, superoxide dismutase, and glutathione peroxidase, and decreased the contents of malondialdehyde and reactive oxygen species and the expression of interleukin-1β, interleukin-6, and tumor necrosis factor-α in the damaged kidneys of the rabbits. In addition, the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream gene heme oxygenase 1 (HO-1) was also inhibited after OTA poisoning, while SeMet activated the Nrf2 signaling pathway and enhanced the expression of Nrf2 and the downstream gene HO-1. In conclusion, SeMet protected against kidney injury caused by OTA in rabbits, and the mechanism may be the activation of the Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Ziqiang Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Yingying Sun
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Hui Xie
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Jiajia Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Xin Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Zhangyu Shi
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Yumei Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, Henan, China.
| |
Collapse
|
10
|
Assar DH, Asa SA, El-Abasy MA, Elbialy ZI, Shukry M, Latif AAE, BinMowyna MN, Althobaiti NA, El-Magd MA. Aspergillus awamori attenuates ochratoxin A-induced renal and cardiac injuries in rabbits by activating the Nrf2/HO-1 signaling pathway and downregulating IL1β, TNFα, and iNOS gene expressions. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:69798-69817. [PMID: 35576029 PMCID: PMC9512883 DOI: 10.1007/s11356-022-20599-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/29/2022] [Indexed: 05/10/2023]
Abstract
Ochratoxin A (OTA) is one of the most dangerous and that pollute agricultural products, inducing a variety of toxic effects in humans and animals. The current study explored the protective effect of different concentrations of Aspergillus awamori (A. awamori) against OTA (0.3 mg/kg diet) induced renal and cardiac damage by exploring its mechanism of action in 60 New Zealand white male rabbits. Dietary supplementation of A. awamori at the selected doses of 50, 100, and 150 mg/kg diet, respectively, for 2 months significantly improved the rabbit's growth performance; modulated the suppressed immune response and restored the altered hematological parameters; reduced the elevated levels of renal injury biomarkers such as urea, creatinine, and alkaline phosphatase; and increased serum total proteins concentrations. Moreover, it also declined enzymatic activities of cardiac injury biomarkers, including AST, LDH, and CK-MB. A. awamori alleviated OTA-induced degenerative and necrotic changes in the kidney and heart of rabbits. Interestingly, A. awamori upregulated Nrf2/OH-1 signaling pathway. Therefore enhanced TAC, CAT, and SOD enzyme activities and reduced OTA-induced oxidative and nitrosative stress by declining iNOS gene expression and consequently lowered MDA and NO levels. In addition to attenuating renal and cardiac inflammation via reducing IL-1β, TNF-α gene expressions in a dose-dependent response. In conclusion,this is the first report to pinpoint that dietary incorporation of A. awamori counteracted OTA-induced renal and cardiac damage by potentiating the rabbit's antioxidant defense system through its potent antioxidant, free radical scavenging, and anti-inflammatory properties in a dose-dependent response. Based on our observations, A. awamori could be utilized as a natural protective agent against ochratoxicosis in rabbits.
Collapse
Affiliation(s)
- Doaa H. Assar
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, 33516 Egypt
| | - Samah Abou Asa
- Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, 33516 Egypt
| | - Moshira A. El-Abasy
- Poultry and Rabbit Diseases Department, Faculty of Veterinary Medicine, Kafr El-Sheikh University, Kafr El-Sheikh, 33516 Egypt
| | - Zizy I. Elbialy
- Fish Processing and Biotechnology Department, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, 33516 Kafr El-Sheikh, Egypt
| | - Mustafa Shukry
- Physiology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, 33516 Egypt
| | - Amera Abd El Latif
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, 33516 Egypt
| | - Mona N. BinMowyna
- College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia
| | - Norah A. Althobaiti
- Biology Department, College of Science and Humanities-Al Quwaiiyah, Shaqra University, Al Quwaiiyah, 19257 Saudi Arabia
| | - Mohammed A. El-Magd
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, 33516 Egypt
| |
Collapse
|
11
|
Yoon JW, Lee SI. Gene expression profiling after ochratoxin A treatment in small intestinal epithelial cells from pigs. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2022; 64:842-853. [PMID: 36287785 PMCID: PMC9574622 DOI: 10.5187/jast.2022.e49] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/09/2022] [Accepted: 06/13/2022] [Indexed: 01/24/2023]
Abstract
Ochratoxin A (OTA) is a well-known mycotoxin that causes disease through the ingestion of contaminated food or feed, for example, in the porcine industry. The intestinal epithelium acts as the first barrier against food contamination. We conducted a study on the exposure of the porcine intestinal epithelium to OTA. We used the intestinal porcine epithelial cell line IPEC-J2 as an in vitro model to evaluate the altered molecular mechanisms following OTA exposure. Gene expression profiling revealed that OTA upregulated 782 genes and downregulated 896, totalling 1678 differentially expressed genes. Furthermore, immunofluorescence, quantitative real-time polymerase chain reaction, and western blotting confirmed that OTA damages the tight junction protein ZO-1. Moreover, OTA activated the expression of inflammatory genes (IL-6, IL-8, IL-10, NF-kB, TLR4, and TNF-α). In summary, this study confirmed that OTA alters various molecular mechanisms and has several adverse effects on IPEC-J2 cells.
Collapse
Affiliation(s)
- Jung Woong Yoon
- Department of Animal Science and
Biotechnology, Kyungpook National University, Sangju 37224,
Korea
| | - Sang In Lee
- Department of Animal Science and
Biotechnology, Kyungpook National University, Sangju 37224,
Korea,Corresponding author: Sang In Lee,
Department of Animal Science and Biotechnology, Kyungpook National University,
Sangju 37224, Korea. Tel: +82-54-530-1943, E-mail:
| |
Collapse
|
12
|
Zhao P, Liu X, Jiang WD, Wu P, Liu Y, Jiang J, Zhang L, Mi HF, Kuang SY, Tang L, Zhou XQ, Feng L. The multiple biotoxicity integrated study in grass carp (Ctenopharyngodon idella) caused by Ochratoxin A: Oxidative damage, apoptosis and immunosuppression. JOURNAL OF HAZARDOUS MATERIALS 2022; 436:129268. [PMID: 35739783 DOI: 10.1016/j.jhazmat.2022.129268] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 05/23/2022] [Accepted: 05/29/2022] [Indexed: 06/15/2023]
Abstract
Ochratoxin A (OTA) is a common hazardous food contaminant that seriously endangers human and animal health. However, limited study is focused on aquatic animal. This research investigated the multiple biotoxicity of OTA on spleen (SP) and head kidney (HK) in grass carp and its related mechanism. Our data showed that, dietary supplemented with OTA above 1209 μg/kg caused histopathological damages by decreasing the number of lymphocytes and necrotizing renal parenchymal cells. Meanwhile, OTA caused oxidative damage and reduced the isoforms mRNAs transcripts of antioxidant enzymes (e.g., GPX1, GPX4, GSTO) partly due to suppressing NF-E2-related factor 2 (Nrf2). OTA triggered apoptosis through mitochondria and death receptor pathway potentially by p38 mitogen-activated protein kinase (p38MAPK) activation. Besides, OTA exacerbated inflammation by down-regulation of anti-inflammatory factor (e.g., IL-10, IL-4) and up-regulations of pro-inflammatory factors (e.g., TNF-α, IL-6), which could be ascribed to signaling meditation of Janus kinase / signal transducer and activator of transcription (JAK/STAT). Additionally, the safe upper limits of OTA were estimated to be 677.6 and 695.08 μg/kg based on the immune-related indexes (C3 contents in the SP and LZ activities in the HK, respectively). Our study has provided a wide insight for toxicological assessment of feed pollutant in aquatic animals.
Collapse
Affiliation(s)
- Piao Zhao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Xin Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China
| | - Jun Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Lu Zhang
- Tongwei Co., Ltd., Chengdu, China, Healthy Aquaculture Key Laboratory of Sichuan Province, Sichuan 610041, China
| | - Hai-Feng Mi
- Tongwei Co., Ltd., Chengdu, China, Healthy Aquaculture Key Laboratory of Sichuan Province, Sichuan 610041, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China.
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan 611130, China.
| |
Collapse
|
13
|
Effects of Dietary Lysophospholipid Inclusion on the Growth Performance, Nutrient Digestibility, Nitrogen Utilization, and Blood Metabolites of Finishing Beef Cattle. Antioxidants (Basel) 2022; 11:antiox11081486. [PMID: 36009204 PMCID: PMC9404894 DOI: 10.3390/antiox11081486] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 12/10/2022] Open
Abstract
This study was conducted to evaluate the effect of dietary supplementation with lysophospholipids (LPLs) on the growth performance, nutrient digestibility, nitrogen utilization, and blood metabolites of finishing beef cattle. In total, 40 Angus beef bulls were blocked for body weight (447 ± 9.64 kg) and age (420 ± 6.1 days) and randomly assigned to one of four treatments (10 beef cattle per treatment): (1) control (CON; basal diet); (2) LLPL (CON supplemented with 0.012% dietary LPL, dry matter (DM) basis); (3) MLPL (CON supplemented with 0.024% dietary LPL, DM basis); and (4) HLPL (CON supplemented with 0.048% dietary LPLs, DM basis). The results showed that dietary supplementation with LPLs linearly increased the average daily gain (p < 0.01), digestibility of DM (p < 0.01), crude protein (p < 0.01), and ether extract (p < 0.01) and decreased the feed conversion ratio (p < 0.01). A linear increase in N retention (p = 0.01) and a decrease in urinary (p = 0.04) and fecal N (p = 0.02) levels were observed with increasing the supplemental doses of LPLs. Bulls fed LPLs showed a linear increase in glutathione peroxidase (p = 0.04) and hepatic lipase (p < 0.01) activity and a decrease in cholesterol (p < 0.01), triglyceride (p < 0.01), and malondialdehyde (p < 0.01) levels. In conclusion, supplementation with LPLs has the potential to improve the growth performance, nutrient digestibility, and antioxidant status of beef cattle.
Collapse
|
14
|
Alleviation of Oral Exposure to Aflatoxin B1-Induced Renal Dysfunction, Oxidative Stress, and Cell Apoptosis in Mice Kidney by Curcumin. Antioxidants (Basel) 2022; 11:antiox11061082. [PMID: 35739979 PMCID: PMC9219944 DOI: 10.3390/antiox11061082] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 05/23/2022] [Accepted: 05/27/2022] [Indexed: 02/06/2023] Open
Abstract
Aflatoxin B1 is a contaminant widely found in food and livestock feed, posing a major threat to human and animal health. Recently, much attention from the pharmaceutical and food industries has been focused on curcumin due to its strong antioxidant capacity. However, the therapeutic impacts and potential mechanisms of curcumin on kidney damage caused by AFB1 are still incomplete. In this study, AFB1 triggered renal injury in mice, as reflected by pathological changes and renal dysfunction. AFB1 induced renal oxidative stress and interfered with the Keap1–Nrf2 pathway and its downstream genes (CAT, SOD1, NQO1, GSS, GCLC, and GCLM), as manifested by elevated oxidative stress metabolites and reduced antioxidant enzymes activities. Additionally, AFB1 was found to increase apoptotic cells percentage in the kidney via the TUNEL assay, along with increased expression of Cyt-c, Bax, cleaved-Caspase-3, Caspase-9, and decreased expression of Bcl-2 at the transcriptional and protein levels; in contrast, for mice given curcumin, there was a significant reversal in kidney coefficient, biochemical parameters, pathological changes, and the expression of genes and proteins involved in oxidative stress and apoptosis. These results indicate that curcumin could antagonize oxidative stress and apoptosis to attenuate AFB1-induced kidney damage.
Collapse
|
15
|
Chansawhang A, Phochantachinda S, Temviriyanukul P, Chantong B. Corticosterone potentiates ochratoxin A-induced microglial activation. Biomol Concepts 2022; 13:230-241. [DOI: 10.1515/bmc-2022-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/10/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
Microglial activation in the central nervous system (CNS) has been associated with brain damage and neurodegenerative disorders. Ochratoxin A (OTA) is a mycotoxin that occurs naturally in food and feed and has been associated with neurotoxicity, while corticosteroids are CNS’ physiological function modulators. This study examined how OTA affected microglia activation and how corticosteroids influenced microglial neuroinflammation. Murine microglial cells (BV-2) were stimulated by OTA, and the potentiation effects on OTA-induced inflammation were determined by corticosterone pre-treatment. Expressions of pro-inflammatory mediators including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), and inducible nitric oxide synthase (iNOS) were determined. Phosphorylation of mitogen-activated protein kinases (MAPKs) was analyzed by western blotting. OTA significantly increased the mRNA expression of IL-6, TNF-α, IL-1β, and iNOS and also elevated IL-6 and NO levels. Corticosterone pre-treatment enhanced the neuroinflammatory response to OTA in a mineralocorticoid receptor (MR)-dependent mechanism, which is associated with increases in extracellular signal-regulated kinase (ERK) and p38 MAPK activation. In response to OTA, microglial cells produced pro-inflammatory cytokines and NO, while corticosterone increased OTA-induced ERK and p38 MAPK phosphorylation via MR. Findings indicated the direct role of OTA in microglia activation and neuroinflammatory response and suggested that low corticosterone concentrations in the brain exacerbated neurodegeneration.
Collapse
Affiliation(s)
- Anchana Chansawhang
- The Center for Veterinary Diagnosis, Faculty of Veterinary Science, Mahidol University , Salaya , Phutthamonthon, Nakhon Pathom 73170 , Thailand
| | - Sataporn Phochantachinda
- Prasu-Arthorn Animal Hospital, Faculty of Veterinary Science, Mahidol University , Salaya , Phutthamonthon, Nakhon Pathom 73170 , Thailand
| | - Piya Temviriyanukul
- Institute of Nutrition, Mahidol University , Salaya , Phutthamonthon, Nakhon Pathom 73170 , Thailand
| | - Boonrat Chantong
- Department of Pre-clinical and Applied Animal Science, Faculty of Veterinary Science, Mahidol University , Salaya , Phutthamonthon, Nakhon Pathom 73170 , Thailand
| |
Collapse
|
16
|
Wu D, Yang C, Yang M, Wu Y, Mao Y, Zhou X, Wang J, Yuan Z, Wu J. Citrinin-Induced Hepatotoxicity in Mice Is Regulated by the Ca 2+/Endoplasmic Reticulum Stress Signaling Pathway. Toxins (Basel) 2022; 14:259. [PMID: 35448868 PMCID: PMC9029441 DOI: 10.3390/toxins14040259] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/23/2022] [Accepted: 04/01/2022] [Indexed: 12/26/2022] Open
Abstract
Citrinin (CTN) is a mycotoxin found in crops and agricultural products and poses a serious threat to human and animal health. The aim of this study is to investigate the hepatotoxicity of CTN in mice and analyze its mechanisms from Ca2+-dependent endoplasmic reticulum (ER) stress perspective. We showed that CTN induced histopathological damage, caused ultrastructural changes in liver cells, and induced abnormal values of biochemical laboratory tests of some liver functions in mice. Treatment with CTN could induce nitric oxide (NO), malondialdehyde (MDA), and reactive oxygen species (ROS) accumulation in mice, accompanied with losses of activities of superoxide dismutase (SOD) and catalase (CAT), levels of glutathione (GSH), and capacities of total antioxidant (T-AOC), resulting in oxidative stress in mice. Furthermore, CTN treatment significantly increased Ca2+ accumulation, upregulated protein expressions of ER stress-mediated apoptosis signal protein (glucose regulated protein 78 (GRP78/BIP), C/EBP-homologous protein (CHOP), Caspase-12, and Caspase-3), and induced hepatocyte apoptosis. These adverse effects were counteracted by 4-phenylbutyric acid (4-PBA), an ER stress inhibitor. In summary, our results showed a possible underlying molecular mechanism for CTN that induced hepatocyte apoptosis in mice by the regulation of the Ca2+/ER stress signaling pathway.
Collapse
Affiliation(s)
- Dongyi Wu
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - Chenglin Yang
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - Mengran Yang
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - You Wu
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - Yan Mao
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - Xinyan Zhou
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - Ji Wang
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - Zhihang Yuan
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - Jing Wu
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (C.Y.); (M.Y.); (Y.W.); (Y.M.); (X.Z.); (J.W.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| |
Collapse
|
17
|
Metabolomic Analysis Reveals the Mechanisms of Hepatotoxicity Induced by Aflatoxin M1 and Ochratoxin A. Toxins (Basel) 2022; 14:toxins14020141. [PMID: 35202168 PMCID: PMC8880135 DOI: 10.3390/toxins14020141] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/15/2021] [Accepted: 12/21/2021] [Indexed: 12/11/2022] Open
Abstract
Aflatoxin M1 (AFM1) is the only toxin with the maximum residue limit in milk, and ochratoxin A (OTA) represents a common toxin in cereals foods. It is common to find the co-occurrence of these two toxins in the environment. However, the interactive effect of these toxins on hepatoxicity and underlying mechanisms is still unclear. The liver and serum metabolomics in mice exposed to individual AFM1 at 3.5 mg/kg b.w., OTA at 3.5 mg/kg b.w., and their combination for 35 days were conducted based on the UPLC-MS method in the present study. Subsequent metabolome on human hepatocellular liver carcinoma (Hep G2) cells was conducted to narrow down the key metabolites. The phenotypic results on liver weight and serum indicators, such as total bilirubin and glutamyltransferase, showed that the combined toxins had more serious adverse effects than an individual one, indicating that the combined AFM1 and OTA displayed synergistic effects on liver damage. Through the metabolic analysis in liver and serum, we found that (i) a synergistic effect was exerted in the combined toxins, because the number of differentially expressed metabolites on combination treatment was higher than the individual toxins, (ii) OTA played a dominant role in the hepatoxicity induced by the combination of AFM1, and OTA and (iii) lysophosphatidylcholines (LysoPCs), more especially, LysoPC (16:1), were identified as the metabolites most affected by AFM1 and OTA. These findings provided a new insight for identifying the potential biomarkers for the hepatoxicity of AFM1 and OTA.
Collapse
|
18
|
Li T, Huang S, Wang J, Yin P, Liu H, Sun C. Alginate oligosaccharides protect against fumonisin B1-induced intestinal damage via promoting gut microbiota homeostasis. Food Res Int 2022; 152:110927. [PMID: 35181098 DOI: 10.1016/j.foodres.2021.110927] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 12/10/2021] [Accepted: 12/20/2021] [Indexed: 11/26/2022]
Abstract
Fumonisin B1 (FB1), one of the most common mycotoxins contaminating feed and food, has been shown to induce intestinal barrier degradation. However, its role on gut microbiota in this process is still unclear. Alginate oligosaccharides (AOS) have been reported to exert their anti-inflammatory and anti-apoptotic function partially via modulation the gut microbiota. However, little is known about the beneficial effect of AOS on gut microbiota upon FB1 exposure. Results show that FB1 degraded intestinal epithelial barrier function as evidenced by increased pathological epithelial cell shedding, reduced the number of goblet cells, and promoted intestinal cell apoptosis. Markedly, FB1 disturbed the cecal and fecal microbiota composition. FB1 increased the level of Lactobacillus and decreased the relative abundance of beneficial microbes. FB1 largely inhibited the production of short chain fatty acids (SCFAs). AOS greatly ameliorated FB1-induced intestinal damage, inflammation, and oxidative stress (eg., T-SOD and MDA). AOS alleviated gut microbial dysbiosis by promoting the growth of beneficial microbes such as Roseburia, Bifidobacterium, and Akkermansia, and increasing SCFAs production upon FB1 exposure. Moreover, the correlation analysis showed that FB1- and AOS-treated gut microbiota alteration is closely associated with the change of intestinal phenotype. We have thus provided a novel insight into the protective role of AOS on FB1-induced gut microbial dysbiosis.
Collapse
Affiliation(s)
- Tiantian Li
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Shimeng Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Jun Wang
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Peng Yin
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Hujun Liu
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Changpo Sun
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China; Standards and Quality Center of National Food and Strategic Reserves Administration, China.
| |
Collapse
|
19
|
Xu D, Yin L, Lin J, Fu H, Peng X, Chang L, Zheng Y, Zhao X, Shu G. Aristolochic Acid I-Induced Hepatotoxicity in Tianfu Broilers Is Associated with Oxidative-Stress-Mediated Apoptosis and Mitochondrial Damage. Animals (Basel) 2021; 11:ani11123437. [PMID: 34944214 PMCID: PMC8698099 DOI: 10.3390/ani11123437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Aristolochic acid (AA) is a component of traditional Chinese herbs and commonly used in the farm poultry industry in China for anti-infection, anti-viral and anti-bacterial treatment. However, long-term and over-exposure of these drugs has been proven to be associated with serious hepatotoxicity, but the mechanism of AA-I-induced hepatotoxicity remains unknown. Therefore, in this study, a subchronic toxicity test was conducted to evaluate the mechanism of AA-I-induced hepatotoxicity in Tianfu broilers. Subchronic exposure to high doses of AA-I in broilers can cause serious hepatotoxicity by breaking the redox balance to form oxidative stress, along with promoting oxidative-stress-mediated apoptosis and mitochondrial damage. In conclusion, AA-I has been found to damage broilers’ livers in high doses. This study provides suggestions for the clinical application of traditional Chinese medicine containing AA-I in the poultry industry. Abstract Aristolochic acid (AA) is a component of traditional Chinese herbs and commonly used for farm animals in China. Over-exposure of AA has been proven to be associated with hepatotoxicity; however, the mechanism of action of AA-I-induced hepatotoxicity remains unknown. In the current study, a subchronic toxicity test was conducted to evaluate the mechanism of AA-induced hepatotoxicity in Tianfu broilers. According to the results, AA-I-induced hepatotoxicity in Tianfu broilers was evidenced by the elevation of liver weight, levels of serum glutamic oxalacetic transaminase (GOT) and glutamic-pyruvic transaminase (GPT). Furthermore, hepatocyte swelling, vesicular degeneration and steatosis were observed. Additionally, AA-I elevated the production of reactive oxygen species (ROS) and induced oxidative stress, which further led to excessive apoptosis, characterized by mitochondrial depolarization, upregulation of Bax, and down-regulation of Bcl-2 expression. In conclusion, the mechanism of AA-I-induced hepatotoxicity was associated with oxidative-stress-mediated apoptosis and mitochondrial damage.
Collapse
Affiliation(s)
- Dan Xu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; (D.X.); (X.Z.)
| | - Lizi Yin
- Department of Basic Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (L.Y.); (J.L.); (H.F.)
| | - Juchun Lin
- Department of Basic Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (L.Y.); (J.L.); (H.F.)
| | - Hualin Fu
- Department of Basic Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (L.Y.); (J.L.); (H.F.)
| | - Xi Peng
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu 611130, China;
| | - Lijen Chang
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Yilei Zheng
- College of Veterinary Medicine, University of Minnesota, Minneapolis, MN 55791, USA;
| | - Xiaoling Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; (D.X.); (X.Z.)
| | - Gang Shu
- Department of Basic Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (L.Y.); (J.L.); (H.F.)
- Correspondence:
| |
Collapse
|
20
|
Hao Y, Xing M, Gu X. Research Progress on Oxidative Stress and Its Nutritional Regulation Strategies in Pigs. Animals (Basel) 2021; 11:1384. [PMID: 34068057 PMCID: PMC8152462 DOI: 10.3390/ani11051384] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress refers to the dramatic increase in the production of free radicals in human and animal bodies or the decrease in the ability to scavenging free radicals, thus breaking the antioxidation-oxidation balance. Various factors can induce oxidative stress in pig production. Oxidative stress has an important effect on pig performance and healthy growth, and has become one of the important factors restricting pig production. Based on the overview of the generation of oxidative stress, its effects on pigs, and signal transduction pathways, this paper discussed the nutritional measures to alleviate oxidative stress in pigs, in order to provide ideas for the nutritional research of anti-oxidative stress in pigs.
Collapse
Affiliation(s)
| | | | - Xianhong Gu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.H.); (M.X.)
| |
Collapse
|
21
|
Li X, Lv Z, Chen J, Nepovimova E, Long M, Wu W, Kuca K. Bacillus amyloliquefaciens B10 can alleviate liver apoptosis and oxidative stress induced by aflatoxin B1. Food Chem Toxicol 2021; 151:112124. [PMID: 33727180 DOI: 10.1016/j.fct.2021.112124] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 12/19/2022]
Abstract
Aflatoxin B1 (AFB1) is a mycotoxin often found in food and livestock feed. It can affect human and animal health and is especially damaging to the liver. This study aims to evaluate whether Bacillus amyloliquefaciens (hereafter referred to as B. amyloliquefaciens) B10 can alleviate the toxic effects of AFB1 and, if so, what mechanism is responsible for its action. Specific pathogen-free (SPF) Kunming mice (5-6 weeks old) were divided into four groups (Control, AFB1, B10 strain, and AFB1 + B10 strain) and conducted continuously via gavage for 28 days. Oxidation indices (MDA, T-AOC, SOD, GSH-Px, and CAT) were then measured using their liver tissues and liver coefficient were calculated. Apoptosis was determined using the TUNEL method. Gene expression was determined for Bax, Bcl-2, BIP, CHOP, JNK, Caspase-12, Caspase-9, and Caspase-3, and protein expression was detected for Bax, Bcl-2, and Caspase-3. Our results showed that AFB1 induced the oxidative damage and apoptosis in the livers of mice. However, for mice given B. amyloliquefaciens B10, the biochemical indices, pathological changes, the expressions of genes and proteins related to oxidative stress and apoptosis were significantly reversed. The results indicate that B. amyloliquefaciens B10 antagonizes oxidative damage and apoptosis induced by AFB1 in the livers of mice. The results of this study are of significance for the future use of this strain to reduce the harm of AFB1 to human health and animal reproductive performance.
Collapse
Affiliation(s)
- Xiaotong Li
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Zhiming Lv
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Jia Chen
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic
| | - Miao Long
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Wenda Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic.
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic.
| |
Collapse
|
22
|
Marin DE, Bulgaru CV, Anghel CA, Pistol GC, Dore MI, Palade ML, Taranu I. Grape Seed Waste Counteracts Aflatoxin B1 Toxicity in Piglet Mesenteric Lymph Nodes. Toxins (Basel) 2020; 12:toxins12120800. [PMID: 33333857 PMCID: PMC7765275 DOI: 10.3390/toxins12120800] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/04/2020] [Accepted: 12/13/2020] [Indexed: 12/20/2022] Open
Abstract
Aflatoxin B1 (AFB1) is a mycotoxin that frequently contaminates cereals and cereal byproducts. This study investigates the effect of AFB1 on the mesenteric lymph nodes (MLNs) of piglets and evaluates if a diet containing grape seed meal (GSM) can counteract the negative effect of AFB1 on inflammation and oxidative stress. Twenty-four weaned piglets were fed the following diets: Control, AFB1 group (320 μg AFB1/kg feed), GSM group (8% GSM), and AFB1 + GSM group (8% GSM + 320 μg AFB1/kg feed) for 30 days. AFB1 has an important antioxidative effect by decreasing the activity of catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GPx) and total antioxidant status. As a result of the exposure to AFB1, an increase of MAP kinases, metalloproteinases, and cytokines, as effectors of an inflammatory response, were observed in the MLNs of intoxicated piglets. GSM induced a reduction of AFB1-induced oxidative stress by increasing the activity of GPx and SOD and by decreasing lipid peroxidation. GSM decreased the inflammatory markers increased by AFB1. These results represent an important and promising way to valorize this waste, which is rich in bioactive compounds, for decreasing AFB1 toxic effects in mesenteric lymph nodes.
Collapse
|
23
|
Shang X, You C, Li X, Yuan L, Jin M, Zhang X. Involvement of 5-HT2 serotonin receptors in cognitive defects induced by aristolochic acid I in mice. Toxicology 2020; 447:152624. [PMID: 33186629 DOI: 10.1016/j.tox.2020.152624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/15/2020] [Accepted: 11/01/2020] [Indexed: 11/17/2022]
Abstract
Aristolochic acids (AAs) are a natural bioactive substance found in Chinese herbs, which are widely used for treating diseases. Many studies have demonstrated that AAs have various pharmacological function, while increasing reports indicated its toxicity. However, the role AAs in cognition remains poorly understood. This study explored the neurotoxic effect of aristolochic acid I (AAI), the most toxic component of the AAs family, on hippocampal synaptic plasticity and spatial cognition in mice. C57BL/6 mice were exposed to 5 mg/kg AAI for 4 weeks. After chronic treatment, AAI considerably increased the level of anxiety and the degree of behavioral despair in mice. Working and reference error rates were higher in the AAI exposed mice than in the control. This was further validated by the molecular docking studies, which AAI might interact with 5-HT2 serotonin receptor (5-HT2AR). Mechanism investigation indicated that AAI triggered inflammation in the hippocampus of mice through increasing the activity of Tnf-α-NF-κB-IL-6 signaling pathway. Conclusively, chronic AAI administration causes inflammation, and it possibly also serves as a potential antagonist of 5-HT2AR to influence the cognition function in C57BL/6 mice.
Collapse
Affiliation(s)
- Xueliang Shang
- School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, PR China.
| | - Congying You
- School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, PR China
| | - Xiang Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, NO. 44 West Culture Road, Ji'nan, 250012, Shandong Province, PR China
| | - Lu Yuan
- School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, PR China
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, PR China.
| | - Xiujun Zhang
- School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, PR China.
| |
Collapse
|
24
|
Zhao H, Jiang N, Han Y, Yang M, Gao P, Xiong X, Xiong S, Zeng L, Xiao Y, Wei L, Li L, Li C, Yang J, Tang C, Xiao L, Liu F, Liu Y, Sun L. Aristolochic acid induces renal fibrosis by arresting proximal tubular cells in G2/M phase mediated by HIF-1α. FASEB J 2020; 34:12599-12614. [PMID: 32706145 DOI: 10.1096/fj.202000949r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/14/2022]
Abstract
Renal tubulointerstitial fibrosis (TIF) is a common pathological feature of aristolochic acid (AA) nephropathy (AAN). G2/M arrest of proximal tubular cells (PTCs) is implicated in renal fibrosis of AAN, but the upstream regulatory molecule remains unknown. Hypoxia inducible factor-1α (HIF-1α) promotes renal fibrosis in kidney disease, but the role of HIF-1α in AAN is unclear. Evidence shows that HIF-1α and p21, a known inducer of cellular G2/M arrest, are closely related to each other. To investigate the role of HIF-1α in renal fibrosis of AAN and its effects on p21 expression and PTCs G2/M arrest, mice with HIF-1α gene knockout PTCs (PT-HIF-1α-KO) were generated, and AAN was induced by AA. In vitro tests were conducted on the human PTCs line HK-2 and primary mouse PTCs. HIF-1α and p21 expression, fibrogenesis, and G2/M arrest of PTCs were determined. Results showed that HIF-1α was upregulated in the kidneys of wild-type (WT) AAN mice, accompanied by p21 upregulation, PTCs G2/M arrest and renal fibrosis, and these alterations were reversed in PT-HIF-1α-KO AAN mice. Similar results were observed in HK-2 cells and were further confirmed in primary PTCs from PT-HIF-1α-KO and WT mice. Inhibiting p21 in HK-2 cells and primary PTCs did not change the expression of HIF-1α, but G2/M arrest and fibrogenesis were reduced. These data indicate that HIF-1α plays a key role in renal fibrosis in AAN by inducing PTCs G2/M arrest modulated through p21. HIF-1α may serve as a potential therapeutic target for AAN.
Collapse
Affiliation(s)
- Hao Zhao
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Na Jiang
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yachun Han
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming Yang
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Peng Gao
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaofen Xiong
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shan Xiong
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lingfeng Zeng
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ying Xiao
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ling Wei
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Li Li
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chenrui Li
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jinfei Yang
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chengyuan Tang
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Li Xiao
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fuyou Liu
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yu Liu
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lin Sun
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
25
|
EFSA Panel on Contaminants in the Food Chain (CONTAM), Schrenk D, Bodin L, Chipman JK, del Mazo J, Grasl‐Kraupp B, Hogstrand C, Hoogenboom L(R, Leblanc J, Nebbia CS, Nielsen E, Ntzani E, Petersen A, Sand S, Schwerdtle T, Vleminckx C, Wallace H, Alexander J, Dall'Asta C, Mally A, Metzler M, Binaglia M, Horváth Z, Steinkellner H, Bignami M. Risk assessment of ochratoxin A in food. EFSA J 2020; 18:e06113. [PMID: 37649524 PMCID: PMC10464718 DOI: 10.2903/j.efsa.2020.6113] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The European Commission asked EFSA to update their 2006 opinion on ochratoxin A (OTA) in food. OTA is produced by fungi of the genus Aspergillus and Penicillium and found as a contaminant in various foods. OTA causes kidney toxicity in different animal species and kidney tumours in rodents. OTA is genotoxic both in vitro and in vivo; however, the mechanisms of genotoxicity are unclear. Direct and indirect genotoxic and non-genotoxic modes of action might each contribute to tumour formation. Since recent studies have raised uncertainty regarding the mode of action for kidney carcinogenicity, it is inappropriate to establish a health-based guidance value (HBGV) and a margin of exposure (MOE) approach was applied. For the characterisation of non-neoplastic effects, a BMDL 10 of 4.73 μg/kg body weight (bw) per day was calculated from kidney lesions observed in pigs. For characterisation of neoplastic effects, a BMDL 10 of 14.5 μg/kg bw per day was calculated from kidney tumours seen in rats. The estimation of chronic dietary exposure resulted in mean and 95th percentile levels ranging from 0.6 to 17.8 and from 2.4 to 51.7 ng/kg bw per day, respectively. Median OTA exposures in breastfed infants ranged from 1.7 to 2.6 ng/kg bw per day, 95th percentile exposures from 5.6 to 8.5 ng/kg bw per day in average/high breast milk consuming infants, respectively. Comparison of exposures with the BMDL 10 based on the non-neoplastic endpoint resulted in MOEs of more than 200 in most consumer groups, indicating a low health concern with the exception of MOEs for high consumers in the younger age groups, indicating a possible health concern. When compared with the BMDL 10 based on the neoplastic endpoint, MOEs were lower than 10,000 for almost all exposure scenarios, including breastfed infants. This would indicate a possible health concern if genotoxicity is direct. Uncertainty in this assessment is high and risk may be overestimated.
Collapse
|
26
|
Yang S, Li L, Yu L, Sun L, Li K, Tong C, Xu W, Cui G, Long M, Li P. Selenium-enriched yeast reduces caecal pathological injuries and intervenes changes of the diversity of caecal microbiota caused by Ochratoxin-A in broilers. Food Chem Toxicol 2020; 137:111139. [DOI: 10.1016/j.fct.2020.111139] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/06/2020] [Accepted: 01/19/2020] [Indexed: 12/11/2022]
|